Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Cancer ; 150(4): 688-704, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34716584

RESUMO

The surface inhibitory receptor NKG2A forms heterodimers with the invariant CD94 chain and is expressed on a subset of activated CD8 T cells. As antibodies to block NKG2A are currently tested in several efficacy trials for different tumor indications, it is important to characterize the NKG2A+ CD8 T cell population in the context of other inhibitory receptors. Here we used a well-controlled culture system to study the kinetics of inhibitory receptor expression. Naïve mouse CD8 T cells were synchronously and repeatedly activated by artificial antigen presenting cells in the presence of the homeostatic cytokine IL-7. The results revealed NKG2A as a late inhibitory receptor, expressed after repeated cognate antigen stimulations. In contrast, the expression of PD-1, TIGIT and LAG-3 was rapidly induced, hours after first contact and subsequently down regulated during each resting phase. This late, but stable expression kinetics of NKG2A was most similar to that of TIM-3 and CD39. Importantly, single-cell transcriptomics of human tumor-infiltrating lymphocytes (TILs) showed indeed that these receptors were often coexpressed by the same CD8 T cell cluster. Furthermore, NKG2A expression was associated with cell division and was promoted by TGF-ß in vitro, although TGF-ß signaling was not necessary in a mouse tumor model in vivo. In summary, our data show that PD-1 reflects recent TCR triggering, but that NKG2A is induced after repeated antigen stimulations and represents a late inhibitory receptor. Together with TIM-3 and CD39, NKG2A might thus mark actively dividing tumor-specific TILs.


Assuntos
Proteínas de Checkpoint Imunológico/fisiologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/fisiologia , Animais , Antígenos CD/fisiologia , Linfócitos T CD8-Positivos/imunologia , Divisão Celular , Receptor Celular 2 do Vírus da Hepatite A/fisiologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores Imunológicos/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Microambiente Tumoral , Proteína do Gene 3 de Ativação de Linfócitos
2.
Front Immunol ; 12: 757836, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34712243

RESUMO

The therapeutic efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is limited by the development of graft-versus-host disease (GVHD). In GVHD, rigorous pre-conditioning regimen resets the immune landscape and inflammatory milieu causing immune dysregulation, characterized by an expansion of alloreactive cells and a reduction in immune regulatory cells. In acute GVHD (aGVHD), the release of damage- and pathogen- associated molecular patterns from damaged tissue caused by the conditioning regimen sets the stage for T cell priming, activation and expansion further exacerbating tissue injury and organ damage, particularly in the gastrointestinal tract. Studies have shown that donor T cells utilize multiple energetic and biosynthetic pathways to mediate GVHD that can be distinct from the pathways used by regulatory T cells for their suppressive function. In chronic GVHD (cGVHD), donor T cells may differentiate into IL-21 producing T follicular helper cells or tissue resident T helper cells that cooperate with germinal center B cells or memory B cells, respectively, to produce allo- and auto-reactive antibodies with subsequent tissue fibrosis. Alternatively, donor T cells can become IFN- γ/IL-17 cytokine expressing T cells that mediate sclerodermatous skin injury. Patients refractory to the first line standard regimens for GVHD treatment have a poor prognosis indicating an urgent need for new therapies to restore the balance between effector and regulatory immune cells while preserving the beneficial graft-versus-tumor effect. Emerging data points toward a role for metabolism in regulating these allo- and auto-immune responses. Here, we will discuss the preclinical and clinical data available on the distinct metabolic demands of acute and chronic GVHD and recent efforts in identifying therapeutic targets using metabolomics. Another dimension of this review will examine the changing microbiome after allo-HSCT and the role of microbial metabolites such as short chain fatty acids and long chain fatty acids on regulating immune responses. Lastly, we will examine the metabolic implications of coinhibitory pathway blockade and cellular therapies in allo-HSCT. In conclusion, greater understanding of metabolic pathways involved in immune cell dysregulation during allo-HSCT may pave the way to provide novel therapies to prevent and treat GVHD.


Assuntos
Doença Enxerto-Hospedeiro/terapia , Metabolômica/tendências , Doença Aguda , Aminoácidos/metabolismo , Doença Crônica , Disbiose/complicações , Disbiose/imunologia , Metabolismo Energético , Ácidos Graxos/fisiologia , Microbioma Gastrointestinal/imunologia , Glutamina/metabolismo , Glicólise , Doença Enxerto-Hospedeiro/metabolismo , Efeito Enxerto vs Tumor , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Proteínas de Checkpoint Imunológico/fisiologia , Imunomodulação , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Metabolômica/métodos , Espécies Reativas de Oxigênio , Subpopulações de Linfócitos T/imunologia , Condicionamento Pré-Transplante/efeitos adversos , Transplante Homólogo/efeitos adversos , Vitaminas/metabolismo
3.
Front Immunol ; 12: 645699, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33968036

RESUMO

Autoimmune diseases, such as multiple sclerosis and type-1 diabetes, are the outcomes of a failure of immune tolerance. Immune tolerance is sustained through interplays between two inter-dependent clusters of immune activities: immune stimulation and immune regulation. The mechanisms of immune regulation are exploited as therapeutic targets for the treatment of autoimmune diseases. One of these mechanisms is immune checkpoints (ICPs). The roles of ICPs in maintaining immune tolerance and hence suppressing autoimmunity were revealed in animal models and validated by the clinical successes of ICP-targeted therapeutics for autoimmune diseases. Recently, these roles were highlighted by the clinical discovery that the blockade of ICPs causes autoimmune disorders. Given the crucial roles of ICPs in immune tolerance, it is plausible to leverage ICPs as a group of therapeutic targets to restore immune tolerance and treat autoimmune diseases. In this review, we first summarize working mechanisms of ICPs, particularly those that have been utilized for therapeutic development. Then, we recount the agents and approaches that were developed to target ICPs and treat autoimmune disorders. These agents take forms of fusion proteins, antibodies, nucleic acids, and cells. We also review and discuss safety information for these therapeutics. We wrap up this review by providing prospects for the development of ICP-targeting therapeutics. In summary, the ever-increasing studies and results of ICP-targeting of therapeutics underscore their tremendous potential to become a powerful class of medicine for autoimmune diseases.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Inibidores de Checkpoint Imunológico/uso terapêutico , Proteínas de Checkpoint Imunológico/fisiologia , Animais , Anticorpos Monoclonais/uso terapêutico , Doenças Autoimunes/etiologia , Doenças Autoimunes/imunologia , Terapia Genética , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Proteínas Virais/fisiologia
4.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-33800752

RESUMO

The members of the B7 family, as immune checkpoint molecules, can substantially regulate immune responses. Since microRNAs (miRs) can regulate gene expression post-transcriptionally, we conducted a scoping review to summarize and discuss the regulatory cross-talk between miRs and new B7 family immune checkpoint molecules, i.e., B7-H3, B7-H4, B7-H5, butyrophilin like 2 (BTNL2), B7-H6, B7-H7, and immunoglobulin like domain containing receptor 2 (ILDR2). The current study was performed using a six-stage methodology structure and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. PubMed, Embase, Scopus, Cochrane, ProQuest, and Google Scholar were systematically searched to obtain the relevant records to 5 November 2020. Two authors independently reviewed the obtained records and extracted the desired data. After quantitative and qualitative analyses, we used bioinformatics approaches to extend our knowledge about the regulatory cross-talk between miRs and the abovementioned B7 family members. Twenty-seven articles were identified that fulfilled the inclusion criteria. Studies with different designs reported gene-miR regulatory axes in various cancer and non-cancer diseases. The regulatory cross-talk between the aforementioned B7 family molecules and miRs might provide valuable insights into the pathogenesis of various human diseases.


Assuntos
Antígenos B7/fisiologia , Proteínas de Checkpoint Imunológico/fisiologia , MicroRNAs/fisiologia , Antígenos B7/genética , Antígenos B7/imunologia , Coagulação Sanguínea , Quimiocinas/fisiologia , Ativação do Complemento , Biologia Computacional/métodos , Doença/genética , Redes Reguladoras de Genes , Humanos , Proteínas de Checkpoint Imunológico/genética , Proteínas de Checkpoint Imunológico/imunologia , MicroRNAs/genética , Transdução de Sinais
5.
Nat Genet ; 53(3): 322-331, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33649593

RESUMO

The expression of inhibitory immune checkpoint molecules, such as programmed death-ligand (PD-L)1, is frequently observed in human cancers and can lead to the suppression of T cell-mediated immune responses. Here, we apply expanded CRISPR-compatible (EC)CITE-seq, a technology that combines pooled CRISPR screens with single-cell mRNA and surface protein measurements, to explore the molecular networks that regulate PD-L1 expression. We also develop a computational framework, mixscape, that substantially improves the signal-to-noise ratio in single-cell perturbation screens by identifying and removing confounding sources of variation. Applying these tools, we identify and validate regulators of PD-L1 and leverage our multimodal data to identify both transcriptional and post-transcriptional modes of regulation. Specifically, we discover that the Kelch-like protein KEAP1 and the transcriptional activator NRF2 mediate the upregulation of PD-L1 after interferon (IFN)-γ stimulation. Our results identify a new mechanism for the regulation of immune checkpoints and present a powerful analytical framework for the analysis of multimodal single-cell perturbation screens.


Assuntos
Antígeno B7-H1/genética , Proteínas de Checkpoint Imunológico/fisiologia , Análise de Célula Única/métodos , Antígeno B7-2/metabolismo , Antígeno B7-H1/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Proteínas Culina/genética , Proteínas Culina/metabolismo , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Receptores de Interferon/genética , Reprodutibilidade dos Testes , Razão Sinal-Ruído , Células THP-1 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
J Hematol Oncol ; 13(1): 83, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32600443

RESUMO

VISTA (V-domain immunoglobulin suppressor of T cell activation) is a well-established immune regulatory receptor. However, pre-clinical investigations indicated more complicated influences of VISTA on cancer immunity than previously recognized. Here, we review the current knowledge on the therapeutic phenotypes and molecular mechanisms that underlie the contradictory roles of VISTA in checking anti-cancer immune responses. Furthermore, we highlight the potential indeterminacy of VISTA-targeted strategies in cancer immunotherapy, with in silico analyses. In fact, VISTA functions like a homeostatic regulator that actively normalizes immune responses. Thus, the regulatory role of VISTA in anti-cancer immunity remains to be fully elucidated.


Assuntos
Antígenos B7/antagonistas & inibidores , Proteínas de Checkpoint Imunológico/fisiologia , Imunoterapia , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/terapia , Subpopulações de Linfócitos T/imunologia , Antígenos B7/química , Antígenos B7/imunologia , Ensaios Clínicos como Assunto , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Proteínas de Checkpoint Imunológico/química , Proteínas de Checkpoint Imunológico/imunologia , Imunomodulação , Linfócitos do Interstício Tumoral/imunologia , Masculino , Proteínas de Neoplasias/química , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Especificidade de Órgãos , Prognóstico , Evasão Tumoral
7.
J Leukoc Biol ; 108(3): 813-824, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32531824

RESUMO

T regulatory (Treg) cells have a major role in the maintenance of immune tolerance against self and foreign antigens through the control of harmful inflammation. Treg cells exert immunosuppressive function by several mechanisms, which can be distinguished as contact dependent or independent. Recently, the secretion of extracellular vesicles (EVs) by Treg cells has been reported as a novel suppressive mechanism capable of modulating immunity in a cell-contact independent and targeted manner, which has been identified in different pathologic scenarios. EVs are cell-derived membranous structures involved in physiologic and pathologic processes through protein, lipid, and genetic material exchange, which allow intercellular communication. In this review, we revise and discuss current knowledge on Treg cells-mediated immune tolerance giving special attention to the production and release of EVs. Multiple studies support that Treg cells-derived EVs represent a refined intercellular exchange device with the capacity of modulating immune responses, thus creating a tolerogenic microenvironment in a cell-free manner. The mechanisms proposed encompass miRNAs-induced gene silencing, the action of surface proteins and the transmission of enzymes. These observations gain relevance by the fact that Treg cells are susceptible to converting into effector T cells after exposition to inflammatory environments. Yet, in contrast to their cells of origin, EVs are unlikely to be modified under inflammatory conditions, highlighting the advantage of their use. Moreover, we speculate in the possibility that Treg cells may contribute to infectious tolerance via vesicle secretion, intervening with CD4+ T cells differentiation and/or stability.


Assuntos
Vesículas Extracelulares/imunologia , Tolerância Imunológica/imunologia , Linfócitos T Reguladores/imunologia , Animais , Subpopulações de Linfócitos B/imunologia , Microambiente Celular , Fatores de Transcrição Forkhead/fisiologia , Inativação Gênica , Humanos , Proteínas de Checkpoint Imunológico/fisiologia , Tolerância Imunológica/genética , Imunoterapia , Inflamação/imunologia , Linfocinas/metabolismo , Camundongos , MicroRNAs/genética , Modelos Imunológicos , Receptores Imunológicos/fisiologia
8.
Cell Res ; 30(4): 285-299, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31974523

RESUMO

Co-inhibitory receptors are important regulators of T-cell function that define the balance between tolerance and autoimmunity. The immune regulatory function of co-inhibitory receptors, including CTLA-4, PD-1, TIM-3, TIGIT, and LAG-3, was first discovered in the setting of autoimmune disease models, in which their blockade or deficiency resulted in induction or exacerbation of the disease. Later on, co-inhibitory receptors on lymphocytes have also been found to influence outcomes in tumor and chronic viral infection settings. These receptors suppress T-cell function in the tumor microenvironment (TME), thereby making the T cells dysfunctional. Based on this observation, blockade of co-inhibitory receptors (also known as checkpoint molecules) has emerged as a successful treatment option for a number of human cancers. However, severe autoimmune-like side effects limit the use of therapeutics that block individual or combinations of co-inhibitory receptors for cancer treatment. In this review we provide an overview of the role of co-inhibitory receptors in autoimmunity and anti-tumor immunity. We then discuss current approaches and future directions to leverage our knowledge of co-inhibitory receptors to target them in tumor immunity without inducing autoimmunity.


Assuntos
Autoimunidade , Proteínas de Checkpoint Imunológico/fisiologia , Neoplasias/metabolismo , Subpopulações de Linfócitos T , Microambiente Tumoral , Animais , Humanos , Imunoterapia , Neoplasias/terapia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia
9.
Front Immunol ; 11: 615317, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33488626

RESUMO

LAG3 is an important immune checkpoint with relevance in cancer, infectious disease and autoimmunity. However, despite LAG3's role in immune exhaustion and the great potential of LAG3 inhibition as treatment, much remains unknown about its biology, particularly its mechanism of action. This review describes the knowns, unknowns and controversies surrounding LAG3. This includes examination of how LAG3 is regulated transcriptionally and post-translationally by endocytosis and proteolytic cleavage. We also discuss the interactions of LAG3 with its ligands and the purpose thereof. Finally, we review LAG3's mechanism of action, including the roles of LAG3 intracellular motifs and the lack of a role for CD4 competition. Overall, understanding the biology of LAG3 can provide greater insight on LAG3 function, which may broaden the appreciation for LAG3's role in disease and potentially aid in the development of targeted therapies.


Assuntos
Antígenos CD/fisiologia , Proteínas de Checkpoint Imunológico/fisiologia , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Motivos de Aminoácidos , Antígenos CD/química , Ensaios Clínicos como Assunto , Células Dendríticas/metabolismo , Endocitose , Regulação da Expressão Gênica , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Proteínas de Checkpoint Imunológico/química , Ligantes , Subpopulações de Linfócitos/metabolismo , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neurônios/metabolismo , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Receptores de Antígenos de Linfócitos T/fisiologia , Transcrição Gênica , Proteína do Gene 3 de Ativação de Linfócitos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...